You are on page 1of 11

Transplant International

REVIEW

Optimized donor management and organ


preservation before kidney transplantation
€mer, Urs Benck& Peter Schnu
Heiko M. Mundt, Benito A. Yard, Bernhard K. Kra € lle

5th Department of Medicine SUMMARY


(Nephrology/Endocrinology/ Kidney transplantation is a major medical improvement for patients with
Rheumatology), Medical Faculty
end-stage renal disease, but organ shortage limits its widespread use. As
Mannheim of the University of
Heidelberg, University Hospital
a consequence, the proportion of grafts procured from extended criteria
Mannheim, Mannheim, Germany
donors (ECD) has increased considerably, but this comes along with
increased rates of delayed graft function (DGF) and a higher incidence
Correspondence of immune-mediated rejection that limits organ and patient survival.
Heiko M. Mundt MD, Theodor- Furthermore, most grafts are derived from brain dead organ donors, but
Kutzer-Ufer 1-3, 68167 Mannheim, the unphysiological state of brain death is associated with significant
Germany. metabolic, hemodynamic, and pro-inflammatory changes, which further
Tel.: 0049 6213835172; compromise patient and graft survival. Thus, donor interventions to
fax: 0049 6213833804; preserve graft quality are fundamental to improve long-term transplanta-
e-mail: heiko.mundt@umm.de tion outcome, but interventions must not harm other potentially trans-
plantable grafts. Several donor pretreatment strategies have provided
encouraging results in animal models, but evidence from human studies
is sparse, as most clinical evidence is derived from single-center or non-
randomized trials. Furthermore, ethical matters have to be considered
especially concerning consent from donors, donor families, and trans-
plant recipients to research in the field of donor treatment. This review
provides an overview of clinically proven and promising preclinical
strategies of donor treatment to optimize long-term results after kidney
transplantation.

Transplant International 2016; 29: 974–984


Key words
brain death, donor management, kidney transplantation, organ preservation

Received: 4 June 2015; Revision requested: 2 July 2015; Accepted: 30 October 2015

or donors aged 50–59 years with two of the following


Introduction
three features: history of hypertension, serum crea-
Kidney transplantation is the treatment of choice in tinine >1.5 mg/dl, or death from cerebrovascular acci-
end-stage renal disease, being superior to dialysis dent. The majority of organs are retrieved from
regarding life expectation and treatment cost [1–3]. donors after brain death (DBD), yet the unphysiologi-
Advances in donor management, standardized surgical cal state of brain death (BD) with metabolic, hemody-
techniques and improved immunosuppression have namic, and pro-inflammatory changes is associated
contributed to enhanced survival rates in the last dec- with impaired graft quality, increased immunogenicity,
ades. Due to the enormous organ shortage, an aug- and compromised patient and graft survival [4,5].
mented use of extended criteria donors (ECD) is Therefore, optimized care of DBDs is fundamental to
needed. ECDs refer to kidney donors aged >60 years maximize the quantity, functional quality and viability

974 ª 2015 Steunstichting ESOT


doi:10.1111/tri.12712
Donor management and organ preservation

of retrievable organs [6]. Improving care for the this approach is almost unfeasible. Furthermore, trans-
potential donor already on the intensive care unit plantations are unpredictable and some recipients
(ICU) has the potential to attenuate irreversible harm remain unknown even after organ procurement. The
to the graft [7]. As organs from ECDs are more prone logistical obstacles are further underlined by the need to
to delayed graft function (DGF) and have a higher proceed quickly with the transplant procedure and the
incidence of immune-mediated rejection, optimized fact that donor treatment should not affect allocation
donor management is of crucial importance [8]. Speci- [13].
fic strategies solely for graft protection can only be Hence, the establishment of institutional review
instituted after death and only with given consent for boards and ethic committees with specialist transplanta-
donation. In line with the danger hypothesis, this tion expertise is advisable to determine the consent pro-
would attenuate the vicious circle of injury and cess subject to the risk of intervention and to decide
increased immunogenicity after transplantation [8]. whether the recipient needs to give informed consent
Experimental studies highlight the benefits of this [9]. Additionally, a safety monitoring board is reason-
approach, but well designed clinical trials studying able, as it can guard each study subject enrolled within
donor pretreatment are sparse [9]. Trials should ide- the clinical trial [14].
ally assess the results of kidney transplantation by
hard outcome data such as patient and graft survival,
Management strategies prior to organ retrieval
but can also consider early parameters such as DGF,
biopsy-proven acute rejection (BPAR), and evolution
Antioxidant agents
of graft function. Here, we discuss current evidence of
specific donor management and organ preservation
Use of recombinant human superoxide dismutase (rh-SOD)
strategies to improve these outcome parameters after
kidney transplantation. Throughout the transplantation course, kidneys are
prone to oxidative stress by multiple pre- and post-
transplant conditions, that is organ procurement, cold
Ethical issues of donor research
preservation, and ischemia reperfusion [15]. Via acti-
The potential of improving donor management is com- vation of signaling molecules, such as nuclear factor-
plicated by a myriad of logistical and ethical challenges. kappa B, oxidative stress promotes inflammation
First, consent to donor research is unique, as interven- through the release of reactive oxygen species [16–18].
tion trials include donors, donor families, and organ Rh-SOD is capable of scavenging free oxygen radicals
recipients, but official guidelines for consent in donor- thereby minimizing oxidative stress. About 200 mg rh-
based research are missing [10]. Donor research cannot SOD given at reperfusion did not affect DGF or
truly harm the deceased and donor0 s consent is not leg- recovery of graft function, but significantly reduced
ally required, but some ethic guidelines propose the the total number of acute rejection episodes (32/33.3%
family to give informed consent [11] while others of 96 controls vs. 15/18.5% of 81 rh-SOD; P < 0.027)
demand a surrogate informed consent for participation and severe acute rejections resulting in graft loss (12/
in clinical trials [12]. Others consider the consent from 12.5% of controls vs. 3/3.7% of rh-SOD; P < 0.038).
the donor family as standard to minimize the family’s Treatment with rh-SOD also improved 4-year graft
emotional distress and to maintain public trust in the survival to 74% compared with 52% in controls. The
medical profession [13]. In general, it can be assumed authors hypothesized that protection of endothelial
that donors and donor families consent to any reason- cells from early reperfusion injury would mitigate the
able scientific effort to improve the outcome of donated process of “chronic obliterative rejection arteriosclero-
organs. Second, if donor research could pose a risk to sis” translating in improved long-term graft survival
the recipient, recipient consent depends on the inter- [19,20].
vention-related risks, ranging from absent to high par-
ticularly when investigational drugs or devices are used.
Donor pretreatment with N-acetylcysteine
If required, consent for donor research should ideally
start when patients join the waiting list within the con- Antioxidant molecules are proposed to limit renal ische-
text of an institutionally approved protocol for any clin- mia-reperfusion injury, but so far clinical studies con-
ical trial. Due to the numerous potential interventions sidering donor treatment with these agents are rare.
and transplant centers involved in different countries, Recently, an open-label monocentre trial with 160
Transplant International 2016; 29: 974–984 975
ª 2015 Steunstichting ESOT
Mundt et al.

DBDs failed to show a beneficial effect of N-acetylcys- tion, steroid use decreased immune-mediated attack
teine pretreatment. N-acetylcysteine neither affected and improved graft function [37,38]. However, these
incidence or duration of DGF nor one-year graft sur- findings could not be confirmed in humans, as early
vival [21]. studies failed to show an advantageous effect of donor
treatment with methylprednisolone [39–43]. Few smal-
ler studies on combined use of cyclophosphamide and
Hormonal resuscitation to antagonize unphysiological
methylprednisolone also suggested improved 5-year
effects after BD
graft survival [40,41], but could not be confirmed by
BD is associated with severe hormonal alterations others [42,43].
resulting from failure of the pituitary gland [22]. Conse- Recently, a large randomized double-blind trial (269
quently, the levels of adrenocorticotropic hormone, cor- DBDs) was conducted to investigate the effects of
tisol, vasopressin, insulin, and triiodothyronine methylprednisolone pretreatment in kidney transplanta-
suddenly drop after the occurrence of BD. Different tion. About 1000 mg methylprednisolone 3 h before
studies have replaced these hormonal deficits. organ retrieval significantly ameliorated gene expression
profiles of inflammatory and apoptotic transcripts but
had no effect on incidence and duration of DGF, or
Blood sugar control and insulin
decline of serum creatinine during the first week.
Today most organ donors have received insulin therapy Hence, high-dose methylprednosolone before organ
already before BD, because monitoring of blood glucose retrieval is not recommended at least in kidney trans-
and insulin therapy is well established in ICUs [23,24]. plantation [36]. It was argued that the negative result of
Current guidelines recommend target glucose levels of the study was due to the very short time window
180 mg/dl (9.9 mmol/l) in the critically ill [25], which between study intervention and initiation of cold
were shown to be safer than lower targets [26,27]. A preservation. Nevertheless, preliminary data suggest that
prospective study from the USA indicated that glucose donor treatment with steroids might be beneficial in
levels above 180 mg/dl are associated with lower organ liver and lung transplantation [44–46]. As retrospective
transplantation rates per donor and worsened graft out- registry-based data indicate that hormonal resuscitation
comes. Therefore, targeting glucose levels of ≤180 mg/dl including methylprednisolone increases the yield of
should be included as a management goal in potential transplantable organs per donor, the routine use of ster-
organ donors [28]. oids as part of a combined hormonal resuscitation has
to be discussed.
Administration of thyroid hormones
Administration of low-dose dopamine
Retrospective studies suggest that hormonal resuscita-
tion including triiodothyronine/L-thyroxine could be As a consequence of BD, sympathetic outflow is inter-
advantageous due to improvements of cardiocirculatory rupted and vasodilatation occurs, leading to hemody-
function with less inotropic requirements [29–31]. Nev- namic instability, so that 80–90% of all DBDs need
ertheless, controlled clinical data and pooled analyses on vasoactive support to maintain adequate organ perfu-
thyroid hormone administration did not confirm a sion. Expert opinions differ, which adrenergic agent
reduced requirement of vasoactive agents, a gain in car- should be administered first-line, as clinical studies on
diac output or an increase in the number of organs pro- use of adrenergic agents focusing on graft outcome are
cured [32,33]. In addition, post-transplant kidney sparse and produced conflicting results [47].
function was not improved [34,35]. Presumably, low Dopamine has traditionally been first choice for
triiodothyronine levels after BD reflect severe injury donors with hemodynamic instability [48], but one ret-
rather than a hypothyroid state [32]. rospective study linked the use of dopamine to an
increased incidence of DGF. Yet, in-depth analysis
revealed that this association was presumably con-
Administration of methylprednisolone
founded by severe hypotension periods in these donors
As endogenous cortisol levels decrease early during BD, [49]. Another retrospective study concluded that use of
their supplementation is advocated because their anti- vasopressors reduced the likelihood of immediate allo-
inflammatory properties might reduce the immunologic graft function [50]. This was confirmed by a prospective
activation after BD [36]. In experimental transplanta- cohort study indicating that donor inotropic support is
976 Transplant International 2016; 29: 974–984
ª 2015 Steunstichting ESOT
Donor management and organ preservation

associated with less immediate graft function and [60,61]. In addition, it was shown that dopamine also
poorer renal graft survival [51]. In contrast, a Canadian increases H2S-production by stimulating endogenous
study identified no dopamine use as a determinant of cystathionine-b-synthase, which protects cells from
initial nonfunction in a multivariate analysis and rec- ROS-formation and apoptosis after cold storage upon
ommended low-dose therapy for all donors [52]. An rewarming [62].
intriguing finding from our center was, that donor
dopamine and to a lesser extent norepinephrine were
Administration of desmopressin (1-deamino-8-D-arginine-
associated with less acute rejection after kidney trans-
vasopressin [DDAVP])
plantation and translated in improved graft survival
[53]. A benefit was also shown in a large multicenter About 80–90% of all DBDs develop central diabetes
cohort study of 2415 kidney transplants from 1993 indi- insipidus with profuse polyuria and potentially severe
cating that adrenergic agents improved 4-year graft sur- dehydration. DDAVP promotes fluid re-absorption in
vival in an apparently dose-dependent manner [54]. the collecting duct and decreases the need for large vol-
Like in the Canadian study, donor dopamine was also ume infusions to hemodynamically stabilize the DBD
associated with reduced dialysis requirements after [63]. Early studies with DDAVP did not show any
transplantation, whereas norepinephrine was not [55]. favorable effect on recipients’ outcome after transplanta-
Based on these data, a multicenter randomized con- tion [64,65]. However, in a more recent study, renal
trolled trial was initiated in 2004, which confirmed that transplant recipients from DDAVP treated donors
treatment of DBDs with low-dose dopamine (4 lg/kg/ showed less rejections episodes and lower serum crea-
min) improves immediate graft function after kidney tinine values 1 and 3 years after transplantation [66].
transplantation. The beneficial effect was enhanced for The retrospective analysis of the dopamine multicenter
kidney grafts with prolonged cold ischemic time (CIT) trial confirmed this observation. While DDAVP pre-
exceeding 17 h and translated in improved graft survival treatment had no effect on short-term outcome such as
in this subgroup. Donor dopamine only infrequently DGF, BPAR, or decline of serum creatinine during the
induced adverse events, namely tachycardia (10.0%) and first week post-transplant, it was significantly associated
hypertension (3.3%), that were reversible after dose with improved 2-year graft survival (85.4% vs. 73.6%,
reduction or premature termination of the dopamine log-rank P = 0.003). Subgroup analyses indicated that
infusion [56]. DDAVP was only beneficial if cold storage was short
(below 14 h) or the donor was assigned to dopamine
pretreatment. Exposure to hypoxia during cold preser-
Molecular mechanism of dopamine
vation and shear stress during reperfusion induces exo-
The advantageous effects of dopamine were not medi- cytosis of Weibel-Palade bodies (WPB) releasing various
ated through hemodynamic stabilization, because all pro-inflammatory cytokines. Both a shorter CIT and
donors were similar with respect to blood pressure and dopamine protect the graft’s endothelium from cold
urine production. Protection is believed to result from storage injury. It was hypothesized that DDAVP treat-
the antioxidant properties of the dopamine molecule ment deprives WPB from the intact endothelium of the
[57]. Cellular damage following prolonged CIT is in part graft before its exposure to ischemia/reperfusion injury.
ascribed to oxidative stress. Under cold storage condi- Reduced release of pro-inflammatory cytokines during
tions, accumulation of reactive oxygen species (ROS) transplantation may attenuate inflammation and trans-
leads to an increased release of calcium ions [58]. A plant vasculopathy.
vicious circle is activated, as intracellular calcium home-
ostasis depends on high energy phosphates which main-
Combined hormonal resuscitation
tain the mitochondrial membrane potential. While
synthesis of ATP is decreased under hypothermia, the In a retrospective analysis of the UNOS database, com-
influx of calcium further exhausts ATP. Abundant intra- bined therapy with methylprednisolone, vasopressin,
cellular calcium aggravates mitochondrial damage, with and triiodothyronine/L-thyroxine as “hormonal resusci-
the consequence that the mitochondrial membrane tation” raised organ yield per donor, especially kidneys
potential ultimately breaks down [59]. We have demon- by 7.3% [67] and was associated with improved kidney
strated that dopamine decelerates the deleterious ampli- graft survival after 1 year [68]. As it is impossible to
fication loop of intracellular calcium accumulation and define the role of any single agent, optimal hormonal
subsequent ATP consumption by scavenging of ROS replacement therapy in DBDs yet remains to be estab-
Transplant International 2016; 29: 974–984 977
ª 2015 Steunstichting ESOT
Mundt et al.

in the recipients [72]. However, these findings were not


Table 1. Overview of hormonal replacement and
vasopressor use in organ donors. confirmed by others [73]. Due to competing interests
regarding optimal treatment of multi-organ donors, lung
Hormone/Treatment Recommendation for administration surgeons would perhaps prefer colloid solutions to stabi-
Insulin for blood Achieve glucose levels of ≤180 mg/dl lize circulation, as gas exchange might be improved by
sugar control [28] attenuation of neurogenic pulmonary edema. Neverthe-
Thyroid hormones Not recommended as a single agent less, evidence so far has linked HES to an increased risk of
Increases number of transplantable
death and renal-replacement therapy in ICUs; therefore,
organs per donor within general
“hormonal resuscitation” [67] use of HES is discouraged [74,75] (Table 2).
Methylprednisolone General consideration
Potential benefit in liver and lung
transplantation [44–46] General donor management goals (DMG)
Increases number of transplantable
International procurement organizations have adopted
organs per donor within general
“hormonal resuscitation” [67] critical care endpoints as management goals for donor
DDAVP Brain death induced diabetes insipidus treatment after confirmation of BD. This resulted in an
(diuresis >5 ml/kg/hr with specific increased organ yield as shown by various retrospective
gravity <1005 mg/ml) analyses [77,78]. Recently, a prospective evaluation by
Might improve kidney graft survival UNOS revealed that a limited number of donors only
[69]
achieve target criteria, but efforts to meet DMG are
Potentially fewer acute rejections
and improved creatinine [66] associated with significantly higher rates of trans-
Vasopressors Kidney donors: Low-dose dopamine plantable organs per donor [79] (Table 3). In addition,
(4 lg/kg/min) DGF after kidney transplantation was less common
Reduction of DGF [56] when DMG were met (17% vs. 30%; P = 0.007) [80]
Improved survival if CIT >17 h [56]
indicating that optimized care for DBDs will not only
extend the pool of organs but also improve clinical out-
come after transplantation.
lished. Table 1 gives an overview of clinically proven
therapies from hormonal resuscitation trials.
Therapeutic hypothermia in deceased organ donors
Therapeutic hypothermia has been shown to be a bene-
Fluid replacement therapy
ficial intervention to protect neurologic function of
Hypovolemia with circulatory collapse is a frequent patients with specific types of cardiac arrest or stroke
complication in DBDs due to central diabetes insipidus, [81–83]. Recently, a large prospective trial in deceased
and loss of sympathetic tone [63]. Hence, adequate fluid organ donors (n = 370) showed that mild hypothermia
replacement is essential to prevent acute renal failure. (34–35 °C) significantly reduced the rate of DGF among
Crystalloid solutions such as 0.9% saline or Ringer’s lac- recipients (28% vs. 39%, odds ratio 0.62; P = 0.02)
tate are considered first choice because they have no [84]. Subgroup analysis showed that high-risk donors,
specific side effects, but may rarely increase edema forma- such as ECDs, particularly benefited from hypothermia
tion. Balanced crystalloid solutions are preferable particu- (odds ratio 0.31; 95% CI 0.15–0.68; P = 0.003).
larly if resuscitation of larger fluid volumes is required,
because administration of 0.9% saline may cause hyper-
Organ storage: Static cold storage or
chloremic metabolic acidosis [7]. Crystalloid fluid load-
hypothermic machine perfusion (MP)
ing to a CVP of 8–10 mmHg may be deleterious to lung
function and should be avoided in potential lung donors Transplants from ECDs are more susceptible to cold
[70]. Colloid solutions could be an alternative to avoid storage inflicted injury, which causes higher rates of
interstitial fluid overload, but they are associated with a DGF and increases the risk for graft failure [85,86].
significant risk of anaphylactic reactions. Furthermore, Therefore, optimizing organ preservation to maintain
HES (hydroxyl ethyl starch) should be avoided in kidney organ quality is a crucial factor for transplantation suc-
donors, because osmotic nephrosis-like lesions were cess. Although static cold preservation is used for the
detected in transplanted kidneys after HES treatment majority of organs transplanted [87,88], MP might be
[71], which were associated with elevated creatinine levels more appropriate to maintain graft viability, especially
978 Transplant International 2016; 29: 974–984
ª 2015 Steunstichting ESOT
Donor management and organ preservation

if organs are retrieved from ECDs or donors after car- (91% vs. 87%; P = 0.04) with MP. Interestingly, the
diac death (DCD) [89]. Early retrospective data indicate benefit of MP could be detected in the subgroup of
that MP may reduce DGF and result in a mild benefit DBDs only (91% vs. 86%; P = 0.02), being pronounced
on death-censored graft survival [90,91]. Another retro- in the subgroup of ECDs (86% vs. 76%; P = 0.01) but
spective study including 912 renal allografts revealed not in DCDs [94]. The suggested beneficial effect for
similar results considering DGF, albeit graft survival ECDs could also be shown in 85 ECD kidneys allocated
remained unaffected [92]. in the Eurotransplant Senior Program. MP reduced the
These initial studies were followed by a large prospec- rate of PNF and improved 1-year graft survival of kid-
tive multicenter trial assessing the influence of MP on neys suffering from DGF, however, no benefit on the
DGF [93]. In this study, one kidney of each donor was incidence of DGF was found, possibly due to the short
randomly assigned to MP or to static cold preservation. CIT of 11 h [95]. However, the above study [93,94] has
MP significantly reduced the rate of DGF (20.8% vs. been criticized, as MP reduced DGF only slightly from
26.5%; P = 0.05) and numerically halved primary non- 89/336 to 70/336, that is, a prevention of 19 episodes in
function (PNF) (2.1% vs. 4.8%; P = 0.08). Duration of 336 transplantations (= 5.7%) and may thus not explain
hospital stay and BPAR were unaffected, but MP signifi- a 4% difference in graft survival. Furthermore, in 25
cantly improved 1-year graft survival (94% vs. 90%, donors assigned to MP in whom vascular anatomy was
P = 0.04). The beneficial effect of MP was similar for considered unsuitable the contralateral kidneys was used
standard criteria and for ECDs. Recently, 3-year follow- instead. These kidney pairs were not excluded but ana-
up data indicated superior long-term graft survival lyzed according to the actual preservation technique
and not “intention to treat”. This ambiguous assign-
ment resulted in a higher number of kidneys with vas-
Table 2. Overview of reported risks and disadvantages of cular abnormalities in controls. Also, MP failed in seven
crystalloid and colloid solutions in potential multi-organ
instances. These kidney pairs were excluded from analy-
donors.
sis, although they were transplanted. Failure of the
Crystalloid solutions Colloid solutions pump could have increased DGF risk and exclusion of
Increase of neurogenic Risk of anaphylactic reactions these kidneys might bias results in favor of MP. There-
pulmonary edema in fore, the value of MP in kidney transplantation is still a
potential lung donors matter of debate, also because other studies could not
[70,76] confirm these findings.
Edema formation Induction of osmotic nephrosis-like
The UK multicenter trial which randomly assigned
lesions [71]
Increased risk of death and DCD kidneys either to MP or static cold storage failed to
renal-replacement therapy show any effect on DGF (58% vs. 56%; P = 0.99). Sur-
in ICUs [74,75] prisingly, MP was associated with an increased rate of
BPAR in the first 3 months (22% vs. 7%; P = 0.06), but
this did not influence graft or patient survival after
12 months [96]. Also, MP was associated with lower graft
Table 3. Recommended donor management goals to
raise organ yield per donor, adapted from [79]. survival in a large registry-based study (n = 2202), even
when the analyses were stratified for duration of CIT [87].
United Network for Organ Sharing (UNOS) region 5 donor
Finally, several meta-analyses have been performed to
management goals
summarize the available evidence on MP in different
Central venous pressure 4–10 mmHg
donor types. A meta-analysis including all donor types
Ejection fraction >50% revealed that MP reduced DGF rates, but had no influence
Vasopressors ≤1 and low dose* on PNF, acute rejections, and graft or patient survival
Arterial blood gas pH 7.3–7.45 [97]. Subsequent meta-analyses were restricted to DCDs
PaO2:FiO2 >300 and found that MP reduced DGF, but the incidence of
Serum sodium 135–155 mmol/l
PNF and 1-year graft or patient survival was unaffected
Blood glucose <150 mg/dl
Urine output 0.5–3 ml/kg/h over 4 h [98,99]. Another meta-analysis considering ECDs only
Mean arterial pressure 60–100 mmHg (2374 MP vs. 8716 CS) concluded that MP was superior in
preventing DGF, and increased 1-year graft survival, how-
*Dopamine ≤10 lg/kg/min, phenylephrine ≤60 lg/kg/min,
ever did not affect PNF or patient survival [100]. Meta-
and norepinephrine ≤10 lg/kg/min.
analysis is limited due to study heterogeneity in terms of
Transplant International 2016; 29: 974–984 979
ª 2015 Steunstichting ESOT
Mundt et al.

the pump systems, perfusion pressures, and cold storage etin diminished the expression of pro-inflammatory genes,
solutions examined [100]. In summary, the available evi- decreased the infiltration of polymorphonuclear cells and
dence about the use of MP is controversial [101]. The use restored kidney function after BD [117]. In a large animal
of MP may prove to be beneficial in high-risk grafts from model of donor pigs erythropoietin decreased renal injury,
ECDs or after prolonged CIT [85]. inflammation and improved kidney function after transplan-
tation [118]. Nevertheless, clinical studies using erythropoi-
etin before surgery and/or following transplantation had no
Preclinical and experimental donor treatment
beneficial effect on graft outcome [119,120]. In one trial,
strategies
administration of erythropoietin even increased the risk of
thrombotic events 1 year after transplantation [121].
Ischemic preconditioning (IPC)
Remote IPC is one potential approach to protect kid-
neys from ischemia-reperfusion injury. Although the Carbon monoxide (CO)
underlying mechanism needs to be fully defined [102], Upcoming evidence suggests that application of carbon
the concept of IPC was shown to be beneficial in rodent monoxide in DBDs is another promising approach for
models of kidney transplantation [103–106], but large prevention of ischemia-reperfusion injury [122]. Addition-
animal models failed to confirm these results [107,108]. ally, it was demonstrated in rat transplant models that CO is
Furthermore, remote IPC by occluding the thigh (three capable of diminishing the graft’s immunogenicity, that is
times for 5 min, either in donors or recipients) failed to donor-derived dendritic cells already before transplantation
improve renal function within 72 h after human living [122–124]. Furthermore, CO was shown to inhibit CAN
donor transplantation [109]. Nonetheless, patients are and to improve survival even when the treatment is started
currently recruited into an interventional randomized after diagnosis of CAN [125]. The promising role of CO in
trial to investigate the effect of remote IPC on immedi- transplantation is reviewed in detail elsewhere [126,127].
ate and 1-year graft function (NCT01395719) [9].

Conclusion
Vagus nerve stimulation
As a consequence of organ shortage, grafts from ECDs
Electric stimulation of the vagal nerve in a BD rodent
need to be used to supply the demand of transplantable
decreased transcription of pro-inflammatory genes,
organs. Grafts from ECDs are more susceptible to vari-
reduced monocyte infiltration of the graft, and improved
its function after transplantation [110]. The mechanism
of action is presumably related to restoring vagus nerve Table 4. Human randomized controlled trials of donor
activity, resulting in recovery of the anti-inflammatory management showing a beneficial effect after kidney
reflex. Recently, also a long-term benefit with less chronic transplantation.
allograft nephropathy (CAN) was shown [111].
Effect on outcome after
Specific donor treatment kidney transplantation
Atorvastatin Glucose levels of <180 mg/dl Higher organ transplantation
rate per donor [28]
In an isogeneic transplantation model, atorvastatin pre- Meeting donor management Higher rate of transplantable
vented ischemia-reperfusion injury and improved renal goals organs per donor [79]
function [112]. Atorvastatin may be beneficial by Reduction of DGF [80]
inhibiting aldose reductase, which plays a major role in Low-dose dopamine Reduction of DGF [56]
oxidative stress [113–115], but the benefit of statin (4 lg/kg/min)
Human superoxide Reduced number of acute
treatment could not be shown in animal transplantation
dismutase (rh-SOD) rejections, improved 4-year
models after BD induction [116]. graft survival [19]
Hypothermic machine Reduced rate of DGF,
perfusion Improved 1- and 3-year graft
Erythropoietin survival, especially for ECDs
High-dose erythropoietin provides anti-apoptotic and [93,94]
Therapeutic hypothermia Reduction of DGF, especially
cytoprotective effects and might enhance the graft’s resis- for ECDs [84]
tance to ischemic injury after BD. In a rat model, erythropoi-
980 Transplant International 2016; 29: 974–984
ª 2015 Steunstichting ESOT
Donor management and organ preservation

ous deleterious events occurring during the course of term results. Important additional issues comprise com-
transplantation. To preserve graft viability, optimized peting interests of some organ-specific interventions [on
donor management is increasingly important. Several quality and procurement of other organs from the same
(pre) clinical donor treatment strategies have revealed donor] and ethical considerations, that is informed con-
promising results, demonstrating the great potential of sent from donors and recipients with regard to trials in
specific interventions in the DBD. donor management. Current donation and allocation sys-
According to the available evidence, donor manage- tems should incorporate donor management protocols to
ment goals have been elaborated to increase organ yield optimize results for transplant recipients and be better
per donor. Regarding specific interventions, randomized designed to facilitate further research that can improve
controlled trials indicate that donor pretreatment with the utility of this most precious resource.
low-dose dopamine or the administration of human
superoxide dismutase at time of reperfusion improve
Funding
the outcome after kidney transplantation by scavenging
of ROS. Additionally, as recently shown, mild hypother- The authors have declared no funding.
mia lowers the rate of DGF among recipients, especially
in high-risk donors like ECDs. While optimal organ
Conflicts of interest
storage is still a matter of debate, current data suggest a
beneficial effect by hypothermic machine perfusion, All authors declare that they do not have any conflict of
especially for ECDs (Table 4). interest with this publication.
In general, overall clinical evidence of donor interven-
tions on graft outcome is sparse and mostly lacks long-

REFERENCES
1. Jassal SV, Krahn MD, Naglie G, et al. review of the literature. Transpl Int last research frontier? Liver Transplant
Kidney transplantation in the elderly: a 2006; 19: 947. 2013; 19: 118.
decision analysis. J Am Soc Nephrol 8. Martins PN, Chandraker A, Tullius SG. 15. Nafar M, Sahraei Z, Salamzadeh J,
2003; 14: 187. Modifying graft immunogenicity and Samavat S, Vaziri ND. Oxidative stress
2. Wolfe RA, Ashby VB, Milford EL, Ojo immune response prior to in kidney transplantation: causes,
AO, Ettenger RE, Agodoa LY, et al. transplantation: potential clinical consequences, and potential treatment.
Comparison of mortality in all patients applications of donor and graft Iran J Kidney Dis 2011; 5: 357.
on dialysis, patients on dialysis treatment. Transpl Int 2006; 19: 351. 16. McCord JM. Oxygen-derived free
awaiting transplantation, and recipients 9. de Vries DK, Wijermars LG, Reinders radicals in postischemic tissue injury.
of a first cadaveric transplant. N Engl J ME, Lindeman JH, Schaapherder AF. N Engl J Med 1985; 312: 159.
Med 1999; 341: 1725. Donor pre-treatment in clinical kidney 17. Parks DA, Granger DN. Ischemia-
3. Schnuelle P, Lorenz D, Trede M, Van transplantation: a critical appraisal. reperfusion injury: a radical view.
Der Woude FJ. Impact of renal Clin Transplant 2013; 27: 799. Hepatology 1988; 8: 680.
cadaveric transplantation on survival in 10. Abt PL, Marsh CL, Dunn TB, et al. 18. Ambros JT, Herrero-Fresneda I, Borau
end-stage renal failure: evidence for Challenges to research and innovation OG, Boira JM. Ischemic
reduced mortality risk compared with to optimize deceased donor organ preconditioning in solid organ
hemodialysis during long-term follow- quality and quantity. Am J Transplant transplantation: from experimental to
up. J Am Soc Nephrol 1998; 9: 2135. 2013; 13: 1400. clinics. Transpl Int 2007; 20: 219.
4. Barklin A. Systemic inflammation in 11. Pentz RD, Cohen CB, Wicclair M, 19. Land W, Schneeberger H, Schleibner S,
the brain-dead organ donor. Acta et al. Ethics guidelines for research et al. The beneficial effect of human
Anaesthesiol Scand 2009; 53: 425. with the recently dead. Nat Med 2005; recombinant superoxide dismutase on
5. Floerchinger B, Oberhuber R, Tullius 11: 1145. acute and chronic rejection events in
SG. Effects of brain death on organ 12. Rey MM, Ware LB, Matthay MA, et al. recipients of cadaveric renal
quality and transplant outcome. Informed consent in research to transplants. Transplantation 1994; 57:
Transplant Rev 2012; 26: 54. improve the number and quality of 211.
6. Mascia L, Mastromauro I, Viberti S, deceased donor organs. Crit Care Med 20. Land W. Innate alloimmunity: history
Vincenzi M, Zanello M. Management 2011; 39: 280. and current knowledge. Exp Clin
to optimize organ procurement in 13. Feng S. Donor intervention and organ Transplant 2007; 5: 575.
brain dead donors. Minerva Anestesiol preservation: where is the science and 21. Orban JC, Quintard H, Cassuto E,
2009; 75: 125. what are the obstacles? Am J Jambou P, Samat-Long C, Ichai C.
7. Schnuelle P, Johannes van der Woude Transplant 2010; 10: 1155. Effect of N-acetylcysteine pretreatment
F. Perioperative fluid management in 14. Mone T, Heldens J, Niemann CU. of deceased organ donors on renal
renal transplantation: a narrative Deceased organ donor research: the allograft function: a randomized

Transplant International 2016; 29: 974–984 981


ª 2015 Steunstichting ESOT
Mundt et al.

controlled trial. Transplantation 2015; kidney cadaver donors. J Heart prospective randomized controlled
99: 746. Transplant 1986; 5: 249. trial. Ann Surg 2008; 248: 1042.
22. Ranasinghe AM, Bonser RS. Endocrine 35. Koller J, Wieser C, Gottardis M, et al. 46. Follette DM, Rudich SM, Babcock WD.
changes in brain death and Thyroid hormones and their impact Improved oxygenation and increased
transplantation. Best Pract Res Clin on the hemodynamic and metabolic lung donor recovery with high-dose
Endocrinol Metab 2011; 25: 799. stability of organ donors and on steroid administration after brain
23. Van den Berghe G. Insulin therapy in kidney graft function after death. J Heart Lung Transplant 1998;
the intensive care unit should be transplantation. Transpl Proc 1990; 22: 17: 423.
targeted to maintain blood glucose 355. 47. Bugge JF. Brain death and its
between 4.4 mmol/l and 6.1 mmol/l. 36. Kainz A, Wilflingseder J, Mitterbauer implications for management of the
Diabetologia 2008; 51: 911. C, et al. Steroid pretreatment of organ potential organ donor. Acta
24. Derde S, Vanhorebeek I, Van den donors to prevent postischemic renal Anaesthesiol Scand 2009; 53: 1239.
Berghe G. Insulin treatment in allograft failure: a randomized, 48. Wood KE, Becker BN, McCartney JG,
intensive care patients. Horm Res 2009; controlled trial. Ann Intern Med 2010; D’Alessandro AM, Coursin DB. Care of
71: 2. 153: 222. the potential organ donor. N Engl J
25. Krinsley JS. Effect of an intensive 37. Reutzel-Selke A, Zschockelt T, Denecke Med 2004; 351: 2730.
glucose management protocol on the C, et al. Short-term 49. Ferguson CJ, Hillis AN, Williams JD,
mortality of critically ill adult patients. immunosuppressive treatment of the Griffin PJ, Salaman JR. Calcium-
Mayo Clin Proc 2004; 79: 992. donor ameliorates consequences of channel blockers and other factors
26. Chase JG, Pretty CG, Pfeifer L, et al. ischemia/reperfusion injury and long- influencing delayed function in renal
Organ failure and tight glycemic term graft function in renal allografts allografts. Nephrol Dial Transplant
control in the SPRINT study. Crit Care from older donors. Transplantation 1990; 5: 816.
2010; 14: R154. 2003; 75: 1786. 50. O’Brien EA, Bour SA, Marshall RL,
27. Finfer S, Chittock DR, Su SY, et al. 38. Pratschke J, Kofla G, Wilhelm MJ, Ahsan N, Yang HC. Effect of use of
Intensive versus conventional glucose et al. Improvements in early behavior vasopressors in organ donors on
control in critically ill patients. N Engl of rat kidney allografts after treatment immediate function of renal allografts.
J Med 2009; 360: 1283. of the brain-dead donor. Ann Surg J Transpl Coord 1996; 6: 215.
28. Sally MB, Ewing T, Crutchfield M, 2001; 234: 732. 51. Marshall R, Ahsan N, Dhillon S,
et al. Determining optimal threshold 39. Chatterjee SN, Terasaki PI, Fine S, Holman M, Yang HC. Adverse effect of
for glucose control in organ donors Schulman B, Smith R, Fine RN. donor vasopressor support on
after neurologic determination of Pretreatment of cadaver donors with immediate and one-year kidney
death: a United Network for Organ methylprednisolone in human renal allograft function. Surgery 1996; 120:
Sharing Region 5 Donor Management allografts. Surg Gynecol Obstet 1977; 663; discussion 6.
Goals Workgroup prospective analysis. 145: 729. 52. Sutherland FR, Bloembergen W,
J Trauma Acute Care Surg 2014; 76: 40. Guttmann RD, Morehouse DD, Mohamed M, Ostbye T, Klar N,
62. Meakins JL, Klassen J, Knaack J, Lazarovits AI. Initial nonfunction in
29. Salim A, Vassiliu P, Velmahos GC, Beaudoin JG. Donor pretreatment in cadaveric renal transplantation. Can J
et al. The role of thyroid hormone an unselected series of cadaver renal Surg 1993; 36: 141.
administration in potential organ allografts. Kidney Int Suppl 1978; 8: 53. Schnuelle P, Lorenz D, Mueller A,
donors. Arch Surg 2001; 136: 1377. S99. Trede M, Van Der Woude FJ. Donor
30. Jeevanandam V, Todd B, Regillo T, 41. Zincke H, Woods JE. Donor catecholamine use reduces acute
Hellman S, Eldridge C, McClurken J. pretreatment in cadaver renal allograft rejection and improves graft
Reversal of donor myocardial transplantation. Surg Gynecol Obstet survival after cadaveric renal
dysfunction by triiodothyronine 1977; 145: 183. transplantation. Kidney Int 1999; 56:
replacement therapy. J Heart Lung 42. Jeffery JR, Downs A, Grahame JW, Lye 738.
Transplant 1994; 13: 681; discussion 5- C, Ramsey E, Thomson AE. A 54. Schnuelle P, Berger S, de Boer J,
7. randomized prospective study of Persijn G, van der Woude FJ. Effects of
31. Zuppa AF, Nadkarni V, Davis L, et al. cadaver donor pretreatment in renal catecholamine application to brain-
The effect of a thyroid hormone transplantation. Transplantation 1978; dead donors on graft survival in solid
infusion on vasopressor support in 25: 287. organ transplantation. Transplantation
critically ill children with cessation of 43. Soulillou JP, Baron D, Rouxel A, 2001; 72: 455.
neurologic function. Crit Care Med Guenel J. Steroid-cyclophosphamide 55. Schnuelle P, Yard BA, Braun C, et al.
2004; 32: 2318. pretreatment of kidney allograft Impact of donor dopamine on
32. Macdonald PS, Aneman A, Bhonagiri donors. A control study. Nephron immediate graft function after kidney
D, et al. A systematic review and 1979; 24: 193. transplantation. Am J Transplant 2004;
meta-analysis of clinical trials of 44. Venkateswaran RV, Patchell VB, 4: 419.
thyroid hormone administration to Wilson IC, et al. Early donor 56. Schnuelle P, Gottmann U, Hoeger S,
brain dead potential organ donors. management increases the retrieval rate et al. Effects of donor pretreatment
Crit Care Med 2012; 40: 1635. of lungs for transplantation. Ann with dopamine on graft function after
33. Powner DJ, Hernandez M. A review of Thorac Surg 2008; 85: 278; discussion kidney transplantation: a randomized
thyroid hormone administration 86. controlled trial. JAMA 2009; 302: 1067.
during adult donor care. Prog 45. Kotsch K, Ulrich F, Reutzel-Selke A, 57. Beck G, Brinkkoetter P, Hanusch C,
Transplant 2005; 15: 202. et al. Methylprednisolone therapy in et al. Clinical review:
34. Gifford RR, Weaver AS, Burg JE, deceased donors reduces inflammation immunomodulatory effects of
Romano PJ, Demers LM, Pennock JL. in the donor liver and improves dopamine in general inflammation.
Thyroid hormone levels in heart and outcome after liver transplantation: a Crit Care 2004; 8: 485.

982 Transplant International 2016; 29: 974–984


ª 2015 Steunstichting ESOT
Donor management and organ preservation

58. Ahlenstiel T, Burkhardt G, Kohler H, 70. Pennefather SH, Bullock RE, Dark JH. 82. van der Worp HB, Macleod MR, Bath
Kuhlmann MK. Bioflavonoids The effect of fluid therapy on alveolar PM, et al. EuroHYP-1: European
attenuate renal proximal tubular cell arterial oxygen gradient in brain-dead multicenter, randomized, phase III
injury during cold preservation in organ donors. Transplantation 1993; clinical trial of therapeutic
Euro-Collins and University of 56: 1418. hypothermia plus best medical
Wisconsin solutions. Kidney Int 2003; 71. Legendre C, Thervet E, Page B, treatment vs. best medical treatment
63: 554. Percheron A, Noel LH, Kreis H. alone for acute ischemic stroke. Int J
59. Haddad P, Cabrillac JC, Piche D, Hydroxyethylstarch and osmotic- Stroke 2014; 9: 642.
Musallam L, Huet PM. Changes in nephrosis-like lesions in kidney 83. Nielsen N, Wetterslev J, Cronberg T,
intracellular calcium induced by acute transplantation. Lancet 1993; 342: 248. et al. Targeted temperature
hypothermia in parenchymal, 72. Cittanova ML, Leblanc I, Legendre C, management at 33 degrees C versus 36
endothelial, and Kupffer cells of the rat Mouquet C, Riou B, Coriat P. Effect of degrees C after cardiac arrest. N Engl J
liver. Cryobiology 1999; 39: 69. hydroxyethylstarch in brain-dead Med 2013; 369: 2197.
60. Yard B, Beck G, Schnuelle P, et al. kidney donors on renal function in 84. Niemann CU, Feiner J, Swain S, et al.
Prevention of cold-preservation injury kidney-transplant recipients. Lancet Therapeutic hypothermia in deceased
of cultured endothelial cells by 1996; 348: 1620. organ donors and kidney-graft
catecholamines and related 73. Deman A, Peeters P, Sennesael J. function. N Engl J Med 2015; 373: 405.
compounds. Am J Transplant 2004; 4: Hydroxyethyl starch does not impair 85. Yuan X, Theruvath AJ, Ge X, et al.
22. immediate renal function in kidney Machine perfusion or cold storage in
61. Brinkkoetter PT, Song H, Losel R, transplant recipients: a retrospective, organ transplantation: indication,
et al. Hypothermic injury: the multicentre analysis. Nephrol Dial mechanisms, and future perspectives.
mitochondrial calcium, ATP and ROS Transplant 1999; 14: 1517. Transpl Int 2010; 23: 561.
love-hate triangle out of balance. Cell 74. Myburgh JA, Finfer S, Bellomo R, 86. Port FK, Bragg-Gresham JL, Metzger
Physiol Biochem 2008; 22: 195. et al. Hydroxyethyl starch or saline for RA, et al. Donor characteristics
62. Talaei F, Bouma HR, Van der Graaf fluid resuscitation in intensive care. N associated with reduced graft survival:
AC, Strijkstra AM, Schmidt M, Engl J Med 2012; 367: 1901. an approach to expanding the pool of
Henning RH. Serotonin and dopamine 75. Perner A, Haase N, Guttormsen AB, kidney donors. Transplantation 2002;
protect from hypothermia/rewarming et al. Hydroxyethyl starch 130/0.42 74: 1281.
damage through the CBS/H2S versus Ringer’s acetate in severe sepsis. 87. Opelz G, Dohler B. Multicenter
pathway. PLoS ONE 2011; 6: e22568. N Engl J Med 2012; 367: 124. analysis of kidney preservation.
63. Dictus C, Vienenkoetter B, 76. Busl KM, Bleck TP. Neurogenic Transplantation 2007; 83: 247.
Esmaeilzadeh M, Unterberg A, Ahmadi pulmonary Edema. Crit Care Med 88. Maathuis MH, Leuvenink HG, Ploeg
R. Critical care management of 2015; 43: 1710. RJ. Perspectives in organ preservation.
potential organ donors: our current 77. Franklin GA, Santos AP, Smith JW, Transplantation 2007; 83: 1289.
standard. Clin Transplant 2009; 23 Galbraith S, Harbrecht BG, Garrison 89. Minor T, Paul A. Hypothermic
(Suppl 21): 2. RN. Optimization of donor reconditioning in organ
64. Hirschl MM, Matzner MP, Huber management goals yields increased transplantation. Curr Opin Org
WO, et al. Effect of desmopressin organ use. Am Surg 2010; 76: 587. Transplant 2013; 18: 161.
substitution during organ procurement 78. Hagan ME, McClean D, Falcone CA, 90. Schold JD, Kaplan B, Howard RJ, Reed
on early renal allograft function. Arrington J, Matthews D, Summe C. AI, Foley DP, Meier-Kriesche HU. Are
Nephrol Dial Transplant 1996; 11: 173. Attaining specific donor management we frozen in time? Analysis of the
65. Guesde R, Barrou B, Leblanc I, et al. goals increases number of organs utilization and efficacy of pulsatile
Administration of desmopressin in transplanted per donor: a quality perfusion in renal transplantation. Am
brain-dead donors and renal function improvement project. Prog Transplant J Transplant 2005; 5: 1681.
in kidney recipients. Lancet 1998; 352: 2009; 91. Wight J, Chilcott J, Holmes M, Brewer
1178. 19: 227. N. The clinical and cost-effectiveness of
66. Nijboer WN, Schuurs TA, van der 79. Malinoski DJ, Patel MS, Daly MC, pulsatile machine perfusion versus cold
Hoeven JA, et al. Effect of brain death Oley-Graybill C, Salim A. The impact storage of kidneys for transplantation
on gene expression and tissue of meeting donor management goals retrieved from heart-beating and non-
activation in human donor kidneys. on the number of organs transplanted heart-beating donors. Health Technol
Transplantation 2004; 78: 978. per donor: results from the United Assess 2003; 7: 1.
67. Rosendale JD, Kauffman HM, McBride Network for Organ Sharing Region 5 92. Matsuoka L, Shah T, Aswad S, et al.
MA, et al. Aggressive pharmacologic prospective donor management goals Pulsatile perfusion reduces the
donor management results in more study. Crit Care Med 2012; 40: 2773. incidence of delayed graft function in
transplanted organs. Transplantation 80. Malinoski DJ, Patel MS, Ahmed O, expanded criteria donor kidney
2003; 75: 482. et al. The impact of meeting donor transplantation. Am J Transplant 2006;
68. Novitzky D, Cooper DK, Rosendale management goals on the development 6: 1473.
JD, Kauffman HM. Hormonal therapy of delayed graft function in kidney 93. Moers C, Smits JM, Maathuis MH,
of the brain-dead organ donor: transplant recipients. Am J Transplant et al. Machine perfusion or cold
experimental and clinical studies. 2013; 13: 993. storage in deceased-donor kidney
Transplantation 2006; 82: 1396. 81. Debaty G, Maignan M, Savary D, et al. transplantation. N Engl J Med 2009;
69. Benck U, Gottmann U, Hoeger S, Impact of intra-arrest therapeutic 360: 7.
et al. Donor desmopressin is associated hypothermia in outcomes of 94. Moers C, Pirenne J, Paul A, Ploeg RJ.
with superior graft survival after prehospital cardiac arrest: a Machine perfusion or cold storage in
kidney transplantation. Transplantation randomized controlled trial. Intensive deceased-donor kidney transplantation.
2011; 92: 1252. Care Med 2014; 40: 1832. N Engl J Med 2012; 366: 770.

Transplant International 2016; 29: 974–984 983


ª 2015 Steunstichting ESOT
Mundt et al.

95. Gallinat A, Moers C, Treckmann J, 106. Jia RP, Zhu JG, Wu JP, Xie JJ, Xu LW. than erythropoietin while preserving
et al. Machine perfusion versus cold Experimental study on early protective renal function. Crit Care Med 2010; 38:
storage for the preservation of kidneys effect of ischemic preconditioning on 1155.
from donors >/= 65 years allocated in rat kidney graft. Transpl Proc 2009; 41: 118. Maio R, Sepodes B, Patel NS,
the Eurotransplant Senior Programme. 69. Thiemermann C, Mota-Filipe H, Costa
Nephrol Dial Transplant 2012; 27: 107. Behrends M, Walz MK, Kribben A, P. Erythropoietin preserves the
4458. et al. No protection of the porcine integrity and quality of organs for
96. Watson CJ, Wells AC, Roberts RJ, kidney by ischaemic preconditioning. transplantation after cardiac death.
et al. Cold machine perfusion versus Exp Physiol 2000; 85: 819. Shock 2011; 35: 126.
static cold storage of kidneys donated 108. Kosieradzki M, Ametani M, Southard 119. Martinez F, Kamar N, Pallet N, et al.
after cardiac death: a UK multicenter JH, Mangino MJ. Is ischemic High dose epoetin beta in the first
randomized controlled trial. Am J preconditioning of the kidney clinically weeks following renal transplantation
Transplant 2010; 10: 1991. relevant? Surgery 2003; 133: 81. and delayed graft function: results of
97. O’Callaghan JM, Morgan RD, Knight 109. Chen Y, Zheng H, Wang X, Zhou Z, the Neo-PDGF Study. Am J Transplant
SR, Morris PJ. Systematic review and Luo A, Tian Y. Remote ischemic 2010; 10: 1695.
meta-analysis of hypothermic machine preconditioning fails to improve early 120. Sureshkumar KK, Hussain SM, Ko TY,
perfusion versus static cold storage of renal function of patients undergoing Thai NL, Marcus RJ. Effect of high-
kidney allografts on transplant living-donor renal transplantation: a dose erythropoietin on graft function
outcomes. Br J Surg 2013; 100: 991. randomized controlled trial. after kidney transplantation: a
98. Deng R, Gu G, Wang D, et al. Transplantation 2013; 95: e4. randomized, double-blind clinical trial.
Machine perfusion versus cold storage 110. Hoeger S, Bergstraesser C, Selhorst J, Clin J Am Soc Nephrol 2012; 7: 1498.
of kidneys derived from donation after et al. Modulation of brain dead 121. Aydin Z, Mallat MJ, Schaapherder AF,
cardiac death: a meta-analysis. PLoS induced inflammation by vagus nerve et al. Randomized trial of short-course
ONE 2013; 8: e56368. stimulation. Am J Transplant 2010; 10: high-dose erythropoietin in donation
99. Bathini V, McGregor T, McAlister VC, 477. after cardiac death kidney transplant
Luke PP, Sener A. Renal perfusion 111. Hoeger S, Fontana J, Jarczyk J, et al. recipients. Am J Transplant 2012; 12:
pump vs cold storage for donation Vagal stimulation in brain dead donor 1793.
after cardiac death kidneys: a rats decreases chronic allograft 122. Caumartin Y, Stephen J, Deng JP,
systematic review. J Urol 2013; 189: nephropathy in recipients. Nephrol et al. Carbon monoxide-releasing
2214. Dial Transplant 2014; 29: 544. molecules protect against ischemia-
100. Jiao B, Liu S, Liu H, Cheng D, Cheng 112. Gottmann U, Brinkkoetter PT, Hoeger reperfusion injury during kidney
Y, Liu Y. Hypothermic machine S, et al. Atorvastatin donor transplantation. Kidney Int 2011; 79:
perfusion reduces delayed graft pretreatment prevents ischemia/ 1080.
function and improves one-year graft reperfusion injury in renal 123. Martins PN, Reutzel-Selke A, Jurisch
survival of kidneys from expanded transplantation in rats: possible role A, et al. Induction of carbon monoxide
criteria donors: a meta-analysis. PLoS for aldose-reductase inhibition. in donor animals prior to organ
ONE 2013; 8: e81826. Transplantation 2007; 84: 755. procurement reduces graft
101. Irish WD, Katz E. Cold machine 113. Amano S, Yamagishi S, Kato N, et al. immunogenicity and inhibits chronic
perfusion or static cold storage of Sorbitol dehydrogenase overexpression allograft dysfunction. Transplantation
kidneys: why the debate continues. Am potentiates glucose toxicity to cultured 2006; 82: 938.
J Transplant 2010; 10: 1955. retinal pericytes. Biochem Biophys Res 124. Kotsch K, Martins PN, Klemz R, et al.
102. Gassanov N, Nia AM, Caglayan E, Er Commun 2002; 299: 183. Heme oxygenase-1 ameliorates
F. Remote ischemic preconditioning 114. Chung SS, Ho EC, Lam KS, Chung ischemia/reperfusion injury by
and renoprotection: from myth to a SK. Contribution of polyol pathway to targeting dendritic cell maturation and
novel therapeutic option? J Am Soc diabetes-induced oxidative stress. J Am migration. Antioxid Redox Signal 2007;
Nephrol 2014; 25: 216. Soc Nephrol 2003; 14(8 Suppl 3): S233. 9: 2049.
103. Murry CE, Jennings RB, Reimer KA. 115. Obrosova IG, Van Huysen C, Fathallah 125. Nakao A, Faleo G, Nalesnik MA, Seda-
Preconditioning with ischemia: a delay L, Cao XC, Greene DA, Stevens MJ. Neto J, Kohmoto J, Murase N. Low-
of lethal cell injury in ischemic An aldose reductase inhibitor reverses dose carbon monoxide inhibits
myocardium. Circulation 1986; 74: early diabetes-induced changes in progressive chronic allograft
1124. peripheral nerve function, metabolism, nephropathy and restores renal
104. Fuller TF, Freise CE, Feng S, and antioxidative defense. FASEB J allograft function. Am J Physiol Renal
Niemann CU. Ischemic 2002; 16: 123. Physiol 2009; 297: F19.
preconditioning improves rat kidney 116. Hoeger S, Benck U, Petrov K, et al. 126. Nakao A, Choi AM, Murase N.
graft function after severe ischemia/ Atorvastatin donor pre-treatment in a Protective effect of carbon monoxide
reperfusion injury. Transpl Proc 2005; model of brain death and allogeneic in transplantation. J Cell Mol Med
37: 377. kidney transplantation in rat. Ann 2006; 10: 650.
105. Jia RP, Xie JJ, Luo FY, Zhu JG. Transplant 2012; 17: 79. 127. Nakao A, Toyoda Y. Application of
Ischemic preconditioning improves rat 117. Nijboer WN, Ottens PJ, van Dijk A, carbon monoxide for transplantation.
kidney allograft function after van Goor H, Ploeg RJ, Leuvenink HG. Curr Pharm Biotechnol 2012; 13: 827.
ischemia/reperfusion injury: the role of Donor pretreatment with carbamylated
tumor necrosis factor-alpha. Transpl erythropoietin in a brain death model
Proc 2008; 40: 3316. reduces inflammation more effectively

984 Transplant International 2016; 29: 974–984


ª 2015 Steunstichting ESOT

You might also like